Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Sci Rep ; 13(1): 5111, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36991111

ABSTRACT

A series of epidemiological studies have shown the limited life expectancy of patients suffering from idiopathic normal pressure hydrocephalus (iNPH). In most cases, comorbid medical conditions are the cause of death, rather than iNPH. Though it has also been shown that shunting improves both life quality and lifetime. We sought to investigate the utility of the Charlson comorbidity index (CCI) for improved preoperative risk-benefit assessment of shunt surgery in individual iNPH cases. 208 shunted iNPH cases were prospectively investigated. Two in-person follow up visits at 3 and 12 months assessed postoperative clinical status. The correlation of the age adjusted CCI with survival was investigated over the median observation time of 2.37 years (IQR 1.16-4.15). Kaplan Meier statistics revealed that patients with a CCI score of 0-5 have a 5-year survival rate of 87%, compared to only 55% in patients with CCI > 5. Cox multivariate statistics revealed that the CCI was an independent predictor of survival, while common preoperative iNPH scores (modified Rankin Scale (mRS), gait score, and continence score) are not. As expected, mRS, gait, and continence scores improved during the postoperative follow up period, though relative improvement on any of these was not predicted by baseline CCI. The CCI is an easily applicable preoperative predictor of survival time in shunted iNPH patients. The lack of a correlation between the CCI and functional outcome means that even patients with multiple comorbidities and limited remaining lifetime may appreciate benefit from shunt surgery.


Subject(s)
Hydrocephalus, Normal Pressure , Humans , Infant , Child, Preschool , Treatment Outcome , Hydrocephalus, Normal Pressure/surgery , Cerebrospinal Fluid Shunts , Ventriculoperitoneal Shunt , Comorbidity
3.
Rare Tumors ; 9(1): 6848, 2017 Mar 24.
Article in English | MEDLINE | ID: mdl-28458792

ABSTRACT

Glomus tumors are rare neoplasms that typically occur within the dermis or subcutis of the subungual space. Primary glomus tumors of the thorax are exceedingly uncommon, thus standard-of-care management is lacking. In this report we describe the management of a patient presenting with a symptomatic glomus tumor of the posterior trachea, and provide a comprehensive review including all documented tracheal glomus tumor reports.

4.
Cancer ; 123(6): 1051-1060, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28263385

ABSTRACT

BACKGROUND: Complete tumor resection is the most important predictor of patient survival with non-small cell lung cancer. Methods for intraoperative margin assessment after lung cancer excision are lacking. This study evaluated near-infrared (NIR) intraoperative imaging with a folate-targeted molecular contrast agent (OTL0038) for the localization of primary lung adenocarcinomas, lymph node sampling, and margin assessment. METHODS: Ten dogs with lung cancer underwent either video-assisted thoracoscopic surgery or open thoracotomy and tumor excision after an intravenous injection of OTL0038. Lungs were imaged with an NIR imaging device both in vivo and ex vivo. The wound bed was re-imaged for retained fluorescence suspicious for positive tumor margins. The tumor signal-to-background ratio (SBR) was measured in all cases. Next, 3 human patients were enrolled in a proof-of-principle study. Tumor fluorescence was measured both in situ and ex vivo. RESULTS: All canine tumors fluoresced in situ (mean Fluoptics SBR, 5.2 [range, 2.7-8.1]; mean Karl Storz SBR 1.9 [range, 1.4-2.6]). In addition, the fluorescence was consistent with tumor margins on pathology. Three positive lymph nodes were discovered with NIR imaging. Also, a positive retained tumor margin was discovered upon NIR imaging of the wound bed. Human pulmonary adenocarcinomas were also fluorescent both in situ and ex vivo (mean SBR, > 2.0). CONCLUSIONS: NIR imaging can identify lung cancer in a large-animal model. In addition, NIR imaging can discriminate lymph nodes harboring cancer cells and also bring attention to a positive tumor margin. In humans, pulmonary adenocarcinomas fluoresce after the injection of the targeted contrast agent. Cancer 2017;123:1051-60. © 2016 American Cancer Society.


Subject(s)
Folate Receptors, GPI-Anchored/metabolism , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Molecular Imaging , Optical Imaging , Spectroscopy, Near-Infrared , Aged , Animals , Contrast Media , Disease Models, Animal , Dogs , Female , Fluorescent Dyes , Humans , Intraoperative Care , Lung Neoplasms/surgery , Lymph Nodes/pathology , Male , Middle Aged , Molecular Imaging/methods , Neoplasm Staging , Optical Imaging/methods , Signal-To-Noise Ratio , Spectroscopy, Near-Infrared/methods , Tomography, X-Ray Computed
5.
Ann Thorac Surg ; 103(2): 390-398, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27793401

ABSTRACT

BACKGROUND: Pulmonary metastasectomy is widely accepted for many tumor types because it may prolong survival and potentially cure some patients. However, intraoperative localization of pulmonary metastases can be technically challenging. We propose that intraoperative near-infrared (NIR) molecular imaging can be used as an adjunct during disease localization. METHODS: We inoculated 50 C57BL/6 mice with Lewis lung carcinoma (LLC) flank tumors. After flank tumor growth, mice were injected through the tail vein with indocyanine green (ICG) before operation, and intraoperative imaging was used to detect pulmonary metastases. On the basis of these experiments, we enrolled 8 patients undergoing pulmonary metastasectomy into a pilot and feasibility clinical trial. Each patient received intravenous ICG 1 day before operation, followed by wedge or segmental resection. Samples were imaged on the back table with an NIR camera to confirm disease presence and margins. All murine and human tumors and margins were confirmed by pathologic examination. RESULTS: Mice had an average of 4 ± 2 metastatic tumors on both lungs, with an average size of 5.1 mm (interquartile range [IQR] 2.2 mm to 7.6 mm). Overall, 200 of 211 (95%) metastatic deposits were markedly fluorescent, with a mean tumor-to-background ratio (TBR) of 3.4 (IQR 3.1 to 4.1). The remaining tumors had a TBR below 1.5. In the human study, intraoperative NIR imaging identified six of the eight preoperatively localized lesions. Intraoperative back table NIR imaging identified all metastatic lesions, which were confirmed by pathologic examination. The average tumor size was 1.75 ± 1.4 cm, and the mean ex vivo TBR was 3.3 (IQR 3.1 to 3.7). Pathologic examination demonstrated melanoma (n = 4), osteogenic sarcoma (n = 2), renal cell carcinoma (n = 2), chondrosarcoma (n = 1), leiomyosarcoma (n = 1), and colorectal carcinoma (n = 1). CONCLUSIONS: Systemic ICG identifies subcentimeter tumor metastases to the lung in murine models, and this work provides proof of principle in humans. Future research is focused on improving depth of penetration into the lung parenchyma.


Subject(s)
Lung Neoplasms/secondary , Molecular Imaging/methods , Neoplasms, Experimental , Pneumonectomy/methods , Spectroscopy, Near-Infrared/methods , Animals , Feasibility Studies , Female , Humans , Intraoperative Period , Lung Neoplasms/diagnosis , Lung Neoplasms/surgery , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Pilot Projects , Retrospective Studies
7.
Neurosurgery ; 79(6): 856-871, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27741220

ABSTRACT

BACKGROUND: Although real-time localization of gliomas has improved with intraoperative image guidance systems, these tools are limited by brain shift, surgical cavity deformation, and expense. OBJECTIVE: To propose a novel method to perform near-infrared (NIR) imaging during glioma resections based on preclinical and clinical investigations, in order to localize tumors and to potentially identify residual disease. METHODS: Fifteen patients were identified and administered a Food and Drug Administration-approved, NIR contrast agent (Second Window indocyanine green [ICG], 5 mg/kg) before surgical resection. An NIR camera was utilized to localize the tumor before resection and to visualize surgical margins following resection. Neuropathology and magnetic resonance imaging data were used to assess the accuracy and precision of NIR fluorescence in identifying tumor tissue. RESULTS: NIR visualization of 15 gliomas (10 glioblastoma multiforme, 1 anaplastic astrocytoma, 2 low-grade astrocytoma, 1 juvenile pilocytic astrocytoma, and 1 ganglioglioma) was performed 22.7 hours (mean) after intravenous injection of ICG. During surgery, 12 of 15 tumors were visualized with the NIR camera. The mean signal-to-background ratio was 9.5 ± 0.8 and fluorescence was noted through the dura to a maximum parenchymal depth of 13 mm. The best predictor of positive fluorescence was enhancement on T1-weighted imaging; this correlated with signal-to-background ratio (P = .03). Nonenhancing tumors did not demonstrate NIR fluorescence. Using pathology as the gold standard, the technique demonstrated a sensitivity of 98% and specificity of 45% to identify tumor in gadolinium-enhancing specimens (n = 71). CONCLUSION: With the use of Second Window ICG, gadolinium-enhancing tumors can be localized through brain parenchyma intraoperatively. Its utility for margin detection is promising but limited by lower specificity. ABBREVIATIONS: 5-ALA, 5-aminolevulinic acidEPR, enhanced permeability and retentionFDA, Food and Drug AdministrationGBM, glioblastomaICG, indocyanine greenNIR, near-infraredNPV, negative predictive valuePPV, positive predictive valueROC, receiver operating characteristicROI, region of interestSBR, signal-to-background ratioWHO, World Health Organization.


Subject(s)
Brain Neoplasms/diagnostic imaging , Brain Neoplasms/surgery , Glioma/diagnostic imaging , Glioma/surgery , Optical Imaging , Spectroscopy, Near-Infrared , Adult , Aminolevulinic Acid , Coloring Agents , Contrast Media , Female , Fluorescence , Gadolinium , Ganglioglioma/pathology , Humans , Indocyanine Green , Magnetic Resonance Imaging , Male , Middle Aged
8.
Discov Med ; 21(116): 275-81, 2016 04.
Article in English | MEDLINE | ID: mdl-27232513

ABSTRACT

Non-small Cell Lung Cancer (NSCLC) remains a deadly disease despite aggressive treatment protocols which incorporate chemotherapy, radiation and surgery. These traditional approaches have reached a plateau in therapeutic benefit. There is emerging evidence suggesting that immunotherapy can serve as an alternative treatment modality for NSCLC. Our group has nearly two decades of experience involving immuno-gene therapy with Ad.hIFN-α and Ad.hIFN-ß in human mesothelioma trials, and has observed both safety and efficacy in treatment of Thoracic malignancies. We have expanded the scope of our work and have obtained encouraging pre-clinical evidence suggesting a role for immunotherapy as a surgical adjuvant for NSCLC cancers. By combining immunotherapy with surgery, synergistic results have been observed. Based on these observations, we have prepared a Phase I Clinical Trial that pairs Ad.hIFN-α with surgery for patients with resectable NSCLC. Patient enrollment is likely to begin in the Summer of 2016. We hope that this trial will serve as a platform for future trials aimed at pairing immunotherapy with surgery for patients diagnosed with NSCLC.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Genetic Therapy/methods , Interferon-alpha/genetics , Lung Neoplasms/therapy , Neoadjuvant Therapy/methods , Adenoviridae/genetics , Animals , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Clinical Trials as Topic , Genetic Vectors/therapeutic use , Glioma/therapy , Humans , Interferon-beta/genetics , Lung Neoplasms/immunology , Mesothelioma/therapy , Neoplasm Staging , Pleural Neoplasms/therapy , Pneumonectomy , Urinary Bladder Neoplasms/therapy
9.
Oncotarget ; 7(3): 3059-67, 2016 Jan 19.
Article in English | MEDLINE | ID: mdl-26689990

ABSTRACT

Near infrared (NIR) molecular imaging is useful to identify tumor margins during surgery; however, the value of this technology has not been evaluated for tumors that have been pre-treated with chemotherapy. We hypothesized that NIR molecular imaging could locate mediastinal tumor margins in a murine model after neoadjuvant chemotherapy. Flank thymomas were established on mice. Two separate experiments were performed for tumor margin detection. The first experiment compared (i) surgery and (ii) surgery + NIR imaging. The second experiment compared (iii) preoperative chemotherapy + surgery, and (iv) preoperative chemotherapy + surgery + NIR imaging. NIR imaging occurred following systemic injection of indocyanine green. Margins were assessed for residual tumor cells by pathology. NIR imaging was superior at detecting retained tumor cells during surgery compared to standard techniques (surgery alone vs. surgery + NIR imaging, 20% vs. 80%, respectively). Following chemotherapy, the sensitivity of NIR imaging of tumor margins was not significantly altered. The mean in vivo tumor-to-background fluorescence ratio was similar in the treatment-naïve and chemotherapy groups ((p = 0.899): 3.79 ± 0.69 (IQR 3.29 - 4.25) vs. 3.79 ± 0.52 (IQR 3.40 - 4.03)). We conclude that chemotherapy does not affect tumor fluorescence or identification of retained cancer cells at margins.


Subject(s)
Mediastinal Neoplasms/diagnosis , Molecular Imaging/methods , Neoplasm, Residual/diagnosis , Optical Imaging/methods , Spectroscopy, Near-Infrared/methods , Thymoma/diagnosis , Animals , Cell Line, Tumor , Mediastinal Neoplasms/drug therapy , Mediastinal Neoplasms/surgery , Mice , Mice, Inbred C57BL , Neoadjuvant Therapy , Thymoma/drug therapy , Thymoma/surgery
10.
Am J Nucl Med Mol Imaging ; 5(4): 390-400, 2015.
Article in English | MEDLINE | ID: mdl-26269776

ABSTRACT

Surgery is the most effective method to cure patients with solid tumors. New techniques in near-infrared (NIR) cancer imaging are being used to identify surgical margins and residual tumor cells in the wound. Our goal was to determine the optimal time and dose for imaging solid tumors using Indocyanine Green. Syngeneic murine flank tumor models were used to test NIR imaging of ICG at various doses ranging from 0 to 10 mg/kg. Imaging was performed immediately after injection and up to 72 hours later. Biodistribution in the blood and murine organs were quantified by spectroscopy and fluorescence microscopy. Based on these results, a six patient dose titration study was performed. In murine flank tumors, the tumor-to-background ratio (TBR) for ICG at doses less than 5 mg/kg were less than 2 fold at all time points, and the surgeons could not subjectively identify tissue contrast. However, for doses ranging from 5 mg/kg to 10 mg/kg, the TBR ranged from 2.1 to 8.0. The tumor signal was best appreciated at 24 hours and the background was least pronounced after 24 hours. Biodistribution studies in the blood and murine organs revealed excretion through the biliary tree and gastrointestinal tract, with minimal blood fluorescence at the higher doses. A follow up pilot study confirmed that these findings were applicable to lung cancer patients, and tumor was clearly delineated from surrounding normal tissue by NIR imaging. For non-hepatic solid tumors, we found ICG was optimal when dosed at 5 mg/kg and 24 hours before surgery.

11.
J Thorac Cardiovasc Surg ; 150(1): 28-35.e1, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26126457

ABSTRACT

BACKGROUND: More than 80,000 people undergo resection of a pulmonary tumor each year, and the only method to determine if the tumor is malignant is histologic analysis. We propose that a targeted molecular contrast agent could bind lung adenocarcinomas, which could be identified using real-time optical imaging at the time of surgery. METHODS: Fifty patients with a biopsy-proven lung adenocarcinoma were enrolled. Before surgery, patients were systemically administered 0.1 mg/kg of a fluorescent folate receptor alpha (FRα)-targeted molecular contrast agent by intravenous infusion. During surgery, tumors were imaged in situ and ex vivo, after the lung parenchyma was dissected to directly expose the tumor to the imaging system. RESULTS: Tumors ranged from 0.3 to 7.5 cm (mean: 2.6 cm), and 46 of 50 (92%) lung adenocarcinomas were fluorescent. No false uptake occurred, and in 2 cases, intraoperative imaging revealed tumor metastases (3 mm and 6 mm) that were not recognized preoperatively. Four adenocarcinomas were not fluorescent, and immunohistochemistry showed that these adenocarcinomas did not express FRα. Tumor fluorescence was independent of nodule size, uptake of 2-deoxy-2-((18)F)fluoro-D-glucose, histology, and tumor differentiation. Molecular imaging could identify only 7 of the 50 adenocarcinomas in situ in the patient without bisection. The most important predictor of the success of molecular imaging in locating the tumor in situ was the distance of the nodule from the pleural surface. CONCLUSIONS: Intraoperative molecular imaging with a targeted contrast agent can identify lung adenocarcinomas, and this technology is currently useful in patients with subpleural tumors, irrespective of size. With further refinements, this tool may prove useful in locating adenocarcinomas that are deeper in the lung parenchyma, in lymph nodes, and at pleural and resection margins.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/surgery , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Molecular Imaging , Monitoring, Intraoperative , Pneumonectomy , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Female , Fluorescein-5-isothiocyanate , Fluorescent Dyes , Humans , Male , Middle Aged , Pilot Projects
12.
J Biomed Opt ; 20(7): 76002, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26160347

ABSTRACT

Surgery is the most effective method to cure patients with solid tumors, and 50% of all cancer patients undergo resection. Local recurrences are due to tumor cells remaining in the wound, thus we explore near-infrared (NIR) fluorescence spectroscopy and imaging to identify residual cancer cells after surgery. Fifteen canines and two human patients with spontaneously occurring sarcomas underwent intraoperative imaging. During the operation, the wounds were interrogated with NIR fluorescence imaging and spectroscopy. NIR monitoring identified the presence or absence of residual tumor cells after surgery in 14/15 canines with a mean fluorescence signal-to-background ratio (SBR) of ∼16 . Ten animals showed no residual tumor cells in the wound bed (mean SBR<2 , P<0.001 ). None had a local recurrence at >1-year follow-up. In five animals, the mean SBR of the wound was >15 , and histopathology confirmed tumor cells in the postsurgical wound in four/five canines. In the human pilot study, neither patient had residual tumor cells in the wound bed, and both remain disease free at >1.5-year follow up. Intraoperative NIR fluorescence imaging and spectroscopy identifies residual tumor cells in surgical wounds. These observations suggest that NIR imaging techniques may improve tumor resection during cancer operations.


Subject(s)
Image Processing, Computer-Assisted/methods , Neoplasm, Residual/pathology , Optical Imaging/methods , Spectroscopy, Near-Infrared/methods , Adult , Animals , Dogs , Humans , Indocyanine Green , Male , Middle Aged , Pilot Projects , Sarcoma/pathology
13.
Technol Cancer Res Treat ; 14(2): 213-20, 2015 Apr.
Article in English | MEDLINE | ID: mdl-24354756

ABSTRACT

Fluorescence guided surgery (FGS) is a developing field of surgical and oncologic research. Practically, FGS has shown useful applications in urologic surgery, benign biliary surgery, colorectal cancer liver metastasis resection, and ovarian cancer debulking. Most notably in in cancer surgery, FGS allows for the clear delineation of cancerous tissue from benign tissue. FGS requires the utilization of a fluorescent contrast agent and an intraoperative fluorescence imaging device (IFID). Currently available IFIDs are expensive, unable to work with multiple fluorophores, and can be cumbersome. This study aims to describe the development and utility of a small, cost-efficient, and interchangeable IFID made from commercially available components. Extensive research was done to design and construct a light-weight, portable, and cost-effective IFID. We researched the capabilities, size, and cost of several camera types and eventually decided on a near-infrared (NIR) charged couple device (CCD) camera for its overall profile. The small portable interchangeable imager of fluorescence (SPIIF) is a "scout" IFID system for FGS. The main components of the SPIIF are a NIR CCD camera with an articulating light filter. These components and a LED light source with an attached heat sink are mounted on a small metal platform. The system is connected to a laptop by a USB 2.0 cable. Pixielink © software on the laptop runs the system by controlling exposure time, gain, and image capture. After developing the system, we evaluated its utility as an IFID. The system weighs less than two pounds and can cover a large area. Due to its small size, it is easily made sterile by covering it with any sterile plastic sheet. To determine the system's ability to detect fluorescent signal, we used the SPIIF to detect indocyanine green under ex and in-vivo conditions and fluorescein under ex-vivo conditions. We found the SPIIF was able to detect both ICG and fluorescein under different depths of a semi-opaque colloid. Second, we found that a concentration as low as 0.5 g/ml of indocyanine green dissolved in plasma was detectable. Lastly, in a murine and human cancer model, the SPIIF was able to detect indocyanine green signal within tumors and generate a signal-to-background ratio (SBR) of 3.75. This study shows that a low-cost IFID can be made from commercially available parts. Second, this IFID is capable of in and ex-vivo detection of multiple fluorophores without sacrificing its small size or favorable ergonomics.


Subject(s)
Bone Neoplasms/surgery , Carcinoma, Lewis Lung/surgery , Carcinoma, Non-Small-Cell Lung/surgery , Optical Imaging/instrumentation , Osteosarcoma/surgery , Surgery, Computer-Assisted/instrumentation , Animals , Carcinoma, Lewis Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/diagnosis , Cell Line, Tumor , Female , Fluorescein , Fluorescent Dyes , Humans , Indocyanine Green , Male , Mice, Inbred C57BL , Middle Aged , Neoplasm Transplantation , Osteosarcoma/diagnosis
14.
Ann Thorac Surg ; 98(4): 1223-30, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25106680

ABSTRACT

BACKGROUND: Over 80,000 people undergo pulmonary resection for a lung nodule in the United States each year. Small nodules are frequently missed or difficult to find despite preoperative imaging. We hypothesized that near-infrared (NIR) imaging technology could be used to identify and locate lung nodules during surgery. METHODS: We enrolled 18 patients who were diagnosed with a pulmonary nodule that required resection. All patients had a fine-cut 1-mm computed tomography scan preoperatively. The patients were given systemic 5 mg/kg indocyanine green and then underwent an open thoracotomy 24 hours later. The NIR imaging was used to identify the primary nodule and search for additional nodules that were not found by visual inspection or manual palpation of the ipsilateral lung. RESULTS: Manual palpation and visual inspection identified all 18 primary pulmonary nodules and no additional lesions. Intraoperative NIR imaging detected 16 out of the 18 primary nodules. The NIR imaging also identified 5 additional subcentimeter nodules; 3 metastatic adenocarcinomas and 2 metastatic sarcomas. This technology could identify nodules as small as 0.2 cm and as deep as 1.3 cm from the pleural surface. This approach discovered 3 nodules that were in different lobes than the primary tumor. Nodule fluorescence was independent of size, metabolic activity, histology, tumor grade and vascularity. CONCLUSIONS: This is the first-in-human demonstration of identifying pulmonary nodules during thoracic surgery with NIR imaging without a priori knowledge of their location or existence. The NIR imaging can detect pulmonary nodules during lung resections that are poorly visualized on computed tomography and difficult to discriminate on finger palpation.


Subject(s)
Solitary Pulmonary Nodule/diagnosis , Adult , Aged , Female , Humans , Indocyanine Green , Intraoperative Period , Lung/pathology , Male , Microscopy, Fluorescence , Middle Aged , Pneumonectomy , Solitary Pulmonary Nodule/blood supply , Solitary Pulmonary Nodule/pathology
15.
PLoS One ; 9(7): e103342, 2014.
Article in English | MEDLINE | ID: mdl-25072388

ABSTRACT

INTRODUCTION: Defining tumor from non-tumor tissue is one of the major challenges of cancer surgery. Surgeons depend on visual and tactile clues to select which tissues should be removed from a patient. Recently, we and others have hypothesized near-infrared (NIR) imaging can be used during surgery to differentiate tumors from normal tissue. METHODS: We enrolled 8 canines and 5 humans undergoing cancer surgery for NIR imaging. The patients were injected with indocyanine green (ICG), an FDA approved non-receptor specific NIR dye that accumulates in hyperpermeable tissues, 16-24 hours prior to surgery. During surgery, NIR imaging was used to discriminate the tumor from non-tumor tissue. RESULTS: NIR imaging identified all tumors with a mean signal-to-background ratio of 6.7. Optical images were useful during surgery in discriminating normal tissue from cancer. In 3 canine cases and 1 human case, the tissue surrounding the tumor was inflamed due to obstruction of the vascular supply due to mass effect. In these instances, NIR imaging could not distinguish tumor tissue from tissue that was congested, edematous and did not contain cancer. CONCLUSIONS: This study shows that NIR imaging can identify tumors from normal tissues, provides excellent tissue contrast, and it facilitates the resection of tumors. However, in situations where there is significant peritumoral inflammation, NIR imaging with ICG is not helpful. This suggests that non-targeted NIR dyes that accumulate in hyperpermeable tissues will have significant limitations in the future, and receptor-specific NIR dyes may be necessary to overcome this problem.


Subject(s)
Diagnostic Imaging/methods , Neoplasms/diagnosis , Spectroscopy, Near-Infrared/methods , Aged , Animals , Cell Line, Tumor , Disease Models, Animal , Dogs , Female , Humans , Indocyanine Green , Inflammation/diagnosis , Intraoperative Period , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/surgery , Mice , Middle Aged , Neoplasm Staging , Neoplasms/pathology , Neoplasms/surgery , Tumor Burden
16.
BMC Immunol ; 14: 30, 2013 Jul 17.
Article in English | MEDLINE | ID: mdl-23865808

ABSTRACT

BACKGROUND: Transforming growth factor (TGF)-ß is a potent immunosuppressive cytokine necessary for cancer growth. Animal and human studies have shown that pharmacologic inhibition of TGF-ß slows the growth rate of established tumors and occasionally eradicates them altogether. We observed, paradoxically, that inhibiting TGF-ß before exposing animals to tumor cells increases tumor growth kinetics. We hypothesized that TGF-ß is necessary for the anti-tumor effects of cytotoxic CD8+ T lymphocytes (CTLs) during the early stages of tumor initiation. METHODS: BALB/c mice were pretreated with a blocking soluble TGF-ß receptor (sTGF-ßR, TGF-ß-blockade group, n=20) or IgG2a (Control group, n=20) before tumor inoculation. Tumor size was followed for 6 weeks. In vivo lymphocyte assays and depletion experiments were then performed to investigate the immunological basis of our results. Lastly, animals were pretreated with either sTGF-ßR (n=6) or IgG2a (n=6) prior to immunization with an adenoviral vector encoding the human papillomavirus E7 gene (Ad.E7). One week later, flow cytometry was utilized to measure the number of splenic E7-specific CD8+ T cells. RESULTS: Inhibition of TGF-ß before the injection of tumor cells resulted in significantly larger average tumor volumes on days 11, 17, 22, 26 and 32 post tumor-inoculation (p < 0.05). This effect was due to the inhibition of CTLs, as it was not present in mice with severe combined immunodeficiency (SCID) or those depleted of CD8+ T cells. Furthermore, pretreatment with sTGF-ßR inhibited tumor-specific CTL activity in a Winn Assay. Tumors grew to a much larger size when mixed with CD8+ T cells from mice pretreated with sTGF-ßR than when mixed with CD8+ T cells from mice in the control group: 96 mm3 vs. 22.5 mm3, respectively (p < 0.05). In addition, fewer CD8+ T cells were generated in Ad.E7-immunized mice pretreated with sTGF-ßR than in mice from the control group: 0.6% total CD8+ T cells vs. 1.9%, respectively (p < 0.05). CONCLUSIONS: These studies provide the first in vivo evidence that TGF-ß may be necessary for anti-tumor immune responses in certain cancers. This finding has important implications for our understanding of anti-tumor immune responses, the role of TGF-ß in the immune system, and the future development of TGF-ß inhibiting drugs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epitopes/immunology , Transforming Growth Factor beta/antagonists & inhibitors , Animals , B7-2 Antigen/metabolism , Cell Count , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Female , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Humans , Immunization , Immunoglobulin G , Lymph Nodes/drug effects , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphocyte Depletion , Mice , Neoplasm Metastasis , Neoplasms/immunology , Neoplasms/pathology , Papillomavirus E7 Proteins/immunology , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction/immunology , Solubility , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Time Factors , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...